Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
BMC Cancer ; 9: 437, 2009 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-20003390

RESUMO

BACKGROUND: Rhabdoid tumors are rare cancers of early childhood arising in the kidney, central nervous system and other organs. The majority are caused by somatic inactivating mutations or deletions affecting the tumor suppressor locus SMARCB1 [OMIM 601607]. Germ-line SMARCB1 inactivation has been reported in association with rhabdoid tumor, epitheloid sarcoma and familial schwannomatosis, underscoring the importance of accurate mutation screening to ascertain recurrence and transmission risks. We describe a rapid and sensitive diagnostic screening method, using high resolution melting (HRM), for detecting sequence variations in SMARCB1. METHODS: Amplicons, encompassing the nine coding exons of SMARCB1, flanking splice site sequences and the 5' and 3' UTR, were screened by both HRM and direct DNA sequencing to establish the reliability of HRM as a primary mutation screening tool. Reaction conditions were optimized with commercially available HRM mixes. RESULTS: The false negative rate for detecting sequence variants by HRM in our sample series was zero. Nine amplicons out of a total of 140 (6.4%) showed variant melt profiles that were subsequently shown to be false positive. Overall nine distinct pathogenic SMARCB1 mutations were identified in a total of 19 possible rhabdoid tumors. Two tumors had two distinct mutations and two harbored SMARCB1 deletion. Other mutations were nonsense or frame-shifts. The detection sensitivity of the HRM screening method was influenced by both sequence context and specific nucleotide change and varied from 1: 4 to 1:1000 (variant to wild-type DNA). A novel method involving digital HRM, followed by re-sequencing, was used to confirm mutations in tumor specimens containing associated normal tissue. CONCLUSIONS: This is the first report describing SMARCB1 mutation screening using HRM. HRM is a rapid, sensitive and inexpensive screening technology that is likely to be widely adopted in diagnostic laboratories to facilitate whole gene mutation screening.


Assuntos
Proteínas Cromossômicas não Histona/genética , Análise Mutacional de DNA/métodos , Proteínas de Ligação a DNA/genética , Tumor Rabdoide/genética , Fatores de Transcrição/genética , Congelamento , Humanos , Mutação , Proteína SMARCB1 , Sensibilidade e Especificidade
2.
Cancer Res ; 67(5): 2360-5, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17325026

RESUMO

Loss of imprinting at insulin-like growth factor II (IGFII), in association with H19 silencing, has been described previously in a subgroup of Beckwith-Wiedemann syndrome (BWS) patients who have an elevated risk for Wilms' tumor. An equivalent somatic mutation occurs in sporadic Wilms' tumor. We describe a family with overgrowth in three generations and Wilms' tumor in two generations, with paternal inheritance of a cis-duplication at 11p15.5 spanning the BWS IC1 region and including H19, IGFII, INS, and TH. The duplicated region was below the limit of detection by high-resolution karyotyping and fluorescence in situ hybridization, has a predicted minimum size of 400 kb, and was confirmed by genotyping and gene-dosage analysis on a CytoChip comparative genomic hybridization bacterial artificial chromosome array. IGFII is the only known paternally expressed oncogene mapping within the duplicated region and our findings directly implicate IGFII in Wilms' tumorigenesis and add to the mutation spectrum that increases the effective dose of IGFII. Furthermore, this study raises the possibility that sporadic cases of overgrowth and Wilms' tumor, presenting with apparent gain of methylation at IC1, may be explained by submicroscopic paternal duplications. This finding has important implications for determining the transmission risk in these disorders.


Assuntos
Cromossomos Humanos Par 11 , Duplicação Gênica , Genes Ligados ao Cromossomo Y , Crescimento/genética , Neoplasias Renais/genética , Proteínas/genética , Proteínas/fisiologia , Tumor de Wilms/genética , Peso ao Nascer/genética , Família , Feminino , Humanos , Lactente , Fator de Crescimento Insulin-Like II , Masculino , Linhagem
3.
Pathology ; 40(7): 664-70, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18985520

RESUMO

AIMS: To correlate the immunostaining for INI1 protein and mutations in INI1 gene in possible rhabdoid tumours (RT) and atypical teratoid/rhabdoid tumours (AT/RT) seen at the Royal Children's Hospital in the last 10 years, and to study the clinicopathological features of those patients with negative nuclear staining. METHODS: Twenty tumours showing suggestive histological and/or immunohistochemical features of RT and AT/RT were selected. Immunohistochemistry for INI1 and molecular investigations for INI1 mutations were performed. The clinical features, histology and immunohistochemistry in those patients with negative nuclear staining were studied. RESULTS: In seven tumours the nuclei stained uniformly for INI1. In none of these was an INI1 mutation detected. In 13 tumours nuclei showed no staining. In only ten of these was material available for molecular studies. Mutations were detected in nine. In these 13 patients, the primary tumour was in the central nervous system (CNS) in seven, in the soft tissue in three, in the liver in two and in the kidney in one. The age of presentation varied from 19 days to 7 years. Only five tumours showed large areas of rhabdoid cells. Most showed extensive non-diagnostic areas. In two an alternative diagnosis, ependymoma or myoepithelial carcinoma of soft tissue, was initially suggested. All the CNS tumours were positive for EMA, GFAP, and SMA. There were no long term survivors, but an occasional patient showed excellent response to intensive chemotherapy. CONCLUSIONS: In this small series, there is a strong correlation between the loss of INI1 immunostaining and the presence of an INI1 mutation suggesting that the former is a reliable marker for RT and AT/RT in children. As relatively few tumours showed uniform populations of rhabdoid cells, and some showed features suggesting another diagnosis, INI1 staining should be checked in all high grade CNS tumours and malignant extraCNS tumours where the diagnosis is unclear. The prognosis of RT is poor but medium term remission can be achieved in some patients with aggressive treatment.


Assuntos
Biomarcadores Tumorais/genética , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Tumor Rabdoide/genética , Tumor Rabdoide/patologia , Fatores de Transcrição/genética , Southern Blotting , Criança , Pré-Escolar , Análise Mutacional de DNA , Feminino , Humanos , Imuno-Histoquímica , Lactente , Recém-Nascido , Masculino , Mutação , Proteína SMARCB1
4.
Clin Epigenetics ; 10(1): 114, 2018 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-30165906

RESUMO

BACKGROUND: Beckwith-Wiedemann syndrome (BWS) is an imprinting disorder with a population frequency of approximately 1 in 10,000. The most common epigenetic defect in BWS is a loss of methylation (LOM) at the 11p15.5 imprinting centre, KCNQ1OT1 TSS-DMR, and affects 50% of cases. We hypothesised that genetic factors linked to folate metabolism may play a role in BWS predisposition via effects on methylation maintenance at KCNQ1OT1 TSS-DMR. RESULTS: Single nucleotide variants (SNVs) in the folate pathway affecting methylenetetrahydrofolate reductase (MTHFR), methionine synthase reductase (MTRR), 5-methyltetrahydrofolate-homocysteine S-methyltransferase (MTR), cystathionine beta-synthase (CBS) and methionine adenosyltransferase (MAT1A) were examined in 55 BWS patients with KCNQ1OT1 TSS-DMR LOM and in 100 unaffected cases. MTHFR rs1801133: C>T was more prevalent in BWS with KCNQ1OT1 TSS-DMR LOM (p < 0.017); however, the relationship was not significant when the Bonferroni correction for multiple testing was applied (significance, p = 0.0036). None of the remaining 13 SNVs were significantly different in the two populations tested. The DNMT1 locus was screened in 53 BWS cases, and three rare missense variants were identified in each of three patients: rs138841970: C>T, rs150331990: A>G and rs757460628: G>A encoding NP_001124295 p.Arg136Cys, p.His1118Arg and p.Arg1223His, respectively. These variants have population frequencies of less than 1 in 1000 and were absent from 100 control cases. Functional characterization using a hemimethylated DNA trapping assay revealed a reduced methyltransferase activity relative to wild-type DNMT1 for each variant ranging from 40 to 70% reduction in activity. CONCLUSIONS: This study is the first to examine folate pathway genetics in BWS and to identify rare DNMT1 missense variants in affected individuals. Our data suggests that reduced DNMT1 activity could affect maintenance of methylation at KCNQ1OT1 TSS-DMR in some cases of BWS, possibly via a maternal effect in the early embryo. Larger cohort studies are warranted to further interrogate the relationship between impaired MTHFR enzymatic activity attributable to MTHFR rs1801133: C>T, dietary folate intake and BWS.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , DNA (Citosina-5-)-Metiltransferase 1/genética , Metilação de DNA , Ácido Fólico/metabolismo , Mutação de Sentido Incorreto , Síndrome de Beckwith-Wiedemann/metabolismo , Feminino , Impressão Genômica , Células HeLa , Humanos , Masculino , Redes e Vias Metabólicas , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Polimorfismo de Nucleotídeo Único , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética
5.
PLoS One ; 6(12): e29034, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22205991

RESUMO

We report a three generation family with Beckwith Wiedemann syndrome (BWS) in whom we have identified a 330 kb deletion within the KCNQ1 locus, encompassing the 11p15.5 Imprinting Centre II (IC2). The deletion arose on the paternal chromosome in the first generation and was only associated with BWS when transmitted maternally to subsequent generations. The deletion on the maternal chromosome was associated with a lower median level of CDKN1C expression in the peripheral blood of affected individuals when compared to a cohort of unaffected controls (p<0.05), however was not significantly different to the expression levels in BWS cases with loss of methylation (LOM) within IC2 (p<0.78). Moreover the individual with a deletion on the paternal chromosome did not show evidence of elevated CDKN1C expression or features of Russell Silver syndrome. These observations support a model invoking the deletion of enhancer elements required for CDKN1C expression lying within or close to the imprinting centre and importantly extend and validate a single observation from an earlier study. Analysis of 94 cases with IC2 loss of methylation revealed that KCNQ1 deletion is a rare cause of loss of maternal methylation, occurring in only 3% of cases, or in 1.5% of BWS overall.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Cromossomos Humanos Par 11/genética , Inibidor de Quinase Dependente de Ciclina p57/genética , Impressão Genômica/genética , Linhagem , Deleção de Sequência/genética , Metilação de DNA/genética , Feminino , Regulação da Expressão Gênica/genética , Haplótipos/genética , Humanos , Canal de Potássio KCNQ1/deficiência , Canal de Potássio KCNQ1/genética , Masculino
6.
PLoS One ; 4(2): e4482, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19221586

RESUMO

SMARCB1 is deleted in rhabdoid tumor, an aggressive paediatric malignancy affecting the kidney and CNS. We hypothesized that the oncogenic pathway in rhabdoid tumors involved epigenetic silencing of key cell cycle regulators as a consequence of altered chromatin-remodelling, attributable to loss of SMARCB1, and that this hypothesis if proven could provide a biological rationale for testing epigenetic therapies in this disease. We used an inducible expression system to show that the imprinted cell cycle inhibitor CDKN1C is a downstream target for SMARCB1 and is transcriptionally activated by increased histone H3 and H4 acetylation at the promoter. We also show that CDKN1C expression induces cell cycle arrest, CDKN1C knockdown with siRNA is associated with increased proliferation, and is able to compete against the anti-proliferative effect of restored SMARCB1 expression. The histone deacetylase inhibitor (HDACi), Romidepsin, specifically restored CDKN1C expression in rhabdoid tumor cells through promoter histone H3 and H4 acetylation, recapitulating the effect of SMARCB1 on CDKNIC allelic expression, and induced cell cycle arrest in G401 and STM91-01 rhabdoid tumor cell lines. CDKN1C expression was also shown to be generally absent in clinical specimens of rhabdoid tumor, however CDKN1A and CDKN1B expression persisted. Our observations suggest that maintenance of CDKN1C expression plays a critical role in preventing rhabdoid tumor growth. Significantly, we report for the first time, parallels between the molecular pathways of SMARCB1 restoration and Romidepsin treatment, and demonstrate a biological basis for the further exploration of histone deacetylase inhibitors as relevant therapeutic reagents in the treatment of rhabdoid tumor.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Proteínas de Ligação a DNA/metabolismo , Impressão Genômica , Inibidores de Histona Desacetilases , Tumor Rabdoide/metabolismo , Fatores de Transcrição/metabolismo , Acetilação , Animais , Antibióticos Antineoplásicos/metabolismo , Linhagem Celular , Proliferação de Células , Criança , Proteínas Cromossômicas não Histona/genética , Inibidor de Quinase Dependente de Ciclina p57/genética , Proteínas de Ligação a DNA/genética , Depsipeptídeos/metabolismo , Histonas/metabolismo , Humanos , Lactente , Camundongos , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Tumor Rabdoide/genética , Proteína SMARCB1 , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA